You are on page 1of 11

REVIEWS

Regulation of cancer cell metabolism


Rob A. Cairns*, Isaac S. Harris* and Tak W. Mak
Abstract | Interest in the topic of tumour metabolism has waxed and waned over the past
century of cancer research. The early observations of Warburg and his contemporaries
established that there are fundamental differences in the central metabolic pathways
operating in malignant tissue. However, the initial hypotheses that were based on these
observations proved inadequate to explain tumorigenesis, and the oncogene revolution
pushed tumour metabolism to the margins of cancer research. In recent years, interest has
been renewed as it has become clear that many of the signalling pathways that are affected
by genetic mutations and the tumour microenvironment have a profound effect on core
metabolism, making this topic once again one of the most intense areas of research in
cancer biology.

Redox status Over the past 25 years, the oncogene revolution has stim- carbohydrates, proteins, lipids and nucleic acids. Many
Balance of the reduced state ulated research, revealing that the crucial phenotypes that similar alterations are also observed in rapidly prolifer-
versus the oxidized state of a are characteristic of tumour cells result from a host of ating normal cells, in which they represent appropriate
biochemical system. This mutational events that combine to alter multiple signalling responses to physiological growth signals as opposed to
balance is influenced by the
level of reactive oxygen and
pathways. Moreover, high-throughput sequencing data constitutive cell autonomous adaptations4,5. In the case
nitrogen species (ROS and suggest that the mutations leading to tumorigenesis are of cancer cells, these adaptations must be implemented
RNS) relative to the capacity of even more numerous and heterogeneous than previously in the stressful and dynamic microenvironment of the
antioxidant systems to thought1,2. It is now clear that there are thousands of point solid tumour, where concentrations of crucial nutrients
eliminate ROS and RNS.
mutations, translocations, amplifications and deletions such as glucose, glutamine and oxygen are spatially and
that may contribute to cancer development, and that the temporally heterogeneous6. The nature and importance
mutational range can differ even among histopathologi- of metabolic restriction in cancer has often been masked
cally identical tumours. Detailed bioinformatic analyses owing to the use of tissue culture conditions in which
have suggested that cancer-related driver mutations affect both oxygen and nutrients are always in excess.
a dozen or more core signalling pathways and processes The link between cancer and altered metabolism is
responsible for tumorigenesis3. These findings have led not new, as many observations made during the early
to questions about the usefulness of targeting individual period of cancer biology research identified metabolic
signalling molecules as a practical therapeutic strategy. changes as a common feature of cancerous tissues (such
However, it is becoming clear that many key oncogenic as the Warburg effect; discussed below)7. As much of the
signalling pathways converge to adapt tumour cell metab- work in the field to date has focused on rapidly prolif-
olism in order to support their growth and survival. erating tumour models and cells in vitro, it is unclear to
Furthermore, some of these metabolic alterations seem what extent these metabolic changes are important in low-
to be absolutely required for malignant transformation. grade slow growing tumours in which metabolic demands
In view of these fundamental discoveries, we propose that are not as extreme. Future clinical data describing the
alterations to cellular metabolism should be considered a metabolic profiles of human tumours will be required to
The Campbell Family Cancer crucial hallmark of cancer. determine which metabolic alterations are most preva-
Research Institute,
Multiple molecular mechanisms, both intrinsic and lent in specific tumour types. However, despite the lack
610 University Avenue,
Toronto, ON M56 2M9, extrinsic, converge to alter core cellular metabolism of comprehensive clinical data, there has been substantial
Canada. and provide support for the three basic needs of dividing recent progress in understanding the molecular events
*These authors contributed cells: rapid ATP generation to maintain energy status; that regulate some of these metabolic phenotypes.
equally to this work. increased biosynthesis of macromolecules; and tightened
Correspondence to T.W.M.
e-mail:
maintenance of appropriate cellular redox status (FIG. 1). To The Warburg effect
tmak@uhnres.utoronto.ca meet these needs, cancer cells acquire alterations to the In addition to the ATP that is required to maintain nor-
doi:10.1038/nrc2981 metabolism of all four major classes of macromolecules: mal cellular processes, proliferating tumour cells must

nATure revIeWs | CanCer vOluMe 11 | FebruAry 2011 | 85

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

At a glance function, and that aerobic glycolysis is therefore a necessary


adaptation to cope with a lack of ATP generation by oxi-
• Multiple molecular mechanisms, both intrinsic and extrinsic, converge to alter core dative phosphorylation. However, it was later appreci-
cellular metabolism and provide support for the three basic needs of dividing cells: ated that mitochondrial defects are rare9 and that most
rapid ATP generation to maintain energy status; increased biosynthesis of
tumours retain the capacity for oxidative phosphorylation
macromolecules; and tightened maintenance of appropriate cellular redox status.
and consume oxygen at rates similar to those observed in
Metabolic changes are a common feature of cancerous tissues, although it is unclear
to what extent these metabolic changes are important in low-grade slow normal tissues10. In fact, mitochondrial function is crucial
growing tumours. for transformation in some systems11–13. Other explana-
• The best characterized metabolic phenotype observed in tumour cells is the Warburg
tions include the concept that glycolysis has the capacity to
effect, which is a shift from ATP generation through oxidative phosphorylation to ATP generate ATP at a higher rate than oxidative phosphory-
generation through glycolysis, even under normal oxygen concentrations. This effect lation and so would be advantageous as long as glucose
is regulated by the PI3K, hypoxia-indicible factor (HIF), p53, MYC and AMP-activated supplies are not limited. Alternatively, it has been pro-
protein kinase (AMPK)–liver kinase B1 (LKB1) pathways. posed that glycolytic metabolism arises as an adaptation
• Metabolic adaptation in tumours extends beyond the Warburg effect. It is becoming to hypoxic conditions during the early avascular phase of
clear that alterations to metabolism balance the need of the cell for energy with its tumour development, as it allows for ATP production in
equally important need for macromolecular building blocks and maintenance of the absence of oxygen. Adaptation to the resulting acidic
redox balance. To this end, a key molecule produced as a result of altered cancer microenvironment that is caused by excess lactate pro-
metabolism is reduced nicotinamide adenine dinucleotide phosphate (NADPH), duction may further drive the evolution of the glycolytic
which functions as a cofactor and provides reducing power in many enzymatic phenotype14,15. Finally, most recently, it has been proposed
reactions that are crucial for macromolecular biosynthesis. NADPH is also an that aerobic glycolysis provides a biosynthetic advantage
antioxidant and forms part of the defence against reactive oxygen species (ROS)
for tumour cells, and that a high flux of substrate through
that are produced during rapid proliferation.
glycolysis allows for effective shunting of carbon to key
• High levels of ROS can cause damage to macromolecules, which can induce
subsidiary biosynthetic pathways4,5.
senescence and apoptosis. Cells counteract the detrimental effects of ROS by
producing antioxidant molecules, such as reduced glutathione (GSH) and thioredoxin
The reliance of cancer cells on increased glucose
(TRX). Several of these antioxidant systems, including GSH and TRX, rely on the uptake has proved useful for tumour detection and
reducing power of NADPH to maintain their activities. monitoring, with this phenotype serving as the basis for
• In addition to the genetic changes that alter tumour cell metabolism, the abnormal
clinical [18F]fluorodeoxyglucose positron emission tom-
tumour microenvironment — such as hypoxia, pH and low glucose concentrations — ography (FDG–PeT) imaging. FDG–PeT uses a radio-
have a major role in determining the metabolic phenotype of tumour cells. active glucose analogue to detect regions of high glucose
• Mutations in oncogenes and tumour suppressor genes cause alterations to multiple uptake, and has proved highly effective for the identifica-
intracellular signalling pathways that affect tumour cell metabolism and re-engineer tion and monitoring of many tumour types. Accordingly,
it to allow enhanced survival and growth. there is now a substantial body of useful clinical data
regarding the importance of glucose as a fuel for malig-
nancies16–19. Although there have been attempts to block
also generate the energy that is required to support rapid aerobic glycolysis in tumour cells using compounds such
cell division. Furthermore, tumour cells must evade the as 2-deoxyglucose, effective therapeutic strategies have
checkpoint controls that would normally block prolif- not yet been devised. several new therapeutic approaches
eration under the stressful metabolic conditions that are targeting numerous points in the glycolytic process are
characteristic of the abnormal tumour microenviron- currently under evaluation, including the inhibition
ment. Tumour cells reprogramme their metabolic path- of lactate dehydrogenase and the inactivation of the
ways to meet these needs during the process of tumour monocarboxylate transporters that are responsible for
progression. The best characterized metabolic phenotype conveying lactate across the plasma membrane20,21.
observed in tumour cells is the Warburg effect (FIG. 2),
which is a shift from ATP generation through oxidative The PI3K pathway. The PI3K pathway is one of the most
phosphorylation to ATP generation through glycolysis, even commonly altered signalling pathways in human can-
under normal oxygen concentrations7. As a result, unlike cers. This pathway is activated by mutations in tumour
Oxidative phosphorylation most normal cells, many transformed cells derive a sub- suppressor genes, such as PTEN, mutations in the com-
Oxygen-dependent process stantial amount of their energy from aerobic glycolysis, ponents of the PI3K complex itself or by aberrant signal-
coupling the oxidation of converting most incoming glucose to lactate rather than ling from receptor tyrosine kinases22. Once activated, the
macromolecules and the
electron transport chain with
metabolizing it in the mitochondria through oxidative PI3K pathway not only provides strong growth and sur-
ATP synthesis. In eukaryotic phosphorylation7,8. Although ATP production by glyco- vival signals to tumour cells but also has profound effects
cells, it occurs within the lysis can be more rapid than by oxidative phosphorylation, on their metabolism. Indeed, it seems that the integra-
mitochondria and is a source of it is far less efficient in terms of ATP generated per unit tion of growth and proliferation signals with alterations
ROS production.
of glucose consumed. This shift therefore demands that to central metabolism is crucial for the oncogenic effects
Glycolysis tumour cells implement an abnormally high rate of glu- of this signalling pathway 23.
Oxygen-independent cose uptake to meet their increased energy, biosynthesis The best-studied effector downstream of PI3K is
metabolism of glucose and and redox needs. AKT1 (also known as PKb). AKT1 is an important
other sugars into pyruvate to There is some debate about the most important selec- driver of the tumour glycolytic phenotype and stimulates
produce energy in the form of
ATP and intermediate
tive advantage that glycolytic metabolism provides to ATP generation through multiple mechanisms, ensur-
substrates for other metabolic proliferating tumour cells. Initial work focused on the con- ing that cells have the bioenergetic capacity required to
pathways. cept that tumour cells develop defects in mitochondrial respond to growth signals24,25. AKT1 stimulates glycolysis

86 | FebruAry 2011 | vOluMe 11 www.nature.com/reviews/cancer

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

by increasing the expression and membrane transloca- conditions, the HIFα subunits undergo oxygen-dependent
tion of glucose transporters and by phosphorylating key hydroxylation by prolyl hydroxylase enzymes, which
glycolytic enzymes, such as hexokinase and phospho- results in their recognition by von Hippel–lindau
fructokinase 2 (also known as PFKFb3)24,26 (FIG. 2). The tumour suppressor (vHl), an e3 ubiquitin ligase,
increased and prolonged AKT1 signalling that is asso- and subsequent degradation. HIF1α is ubiquitously
ciated with transformation inhibits forkhead box sub- expressed, whereas the expression of HIF2α is restricted
family O (FOXO) transcription factors, resulting in a host to a more limited subset of cell types30. Although these
of complex transcriptional changes that increase glyco- two transcription factors transactivate an overlapping set
lytic capacity 27. AKT1 also activates ectonucleoside tri- of genes, the effects on central metabolism have been bet-
phosphate diphosphohydrolase 5 (enTPD5), an enzyme ter characterized for HIF1, and therefore our discussion
that supports increased protein glycosylation in the is limited to HIF1 specifically.
endoplasmic reticulum and indirectly increases glyco- In addition to its stabilization under hypoxic con-
lysis by creating an ATP hydrolysis cycle28. Finally, AKT1 ditions, HIF1 can also be activated under normoxic
strongly stimulates signalling through the kinase mTOr conditions by oncogenic signalling pathways, including
by phosphorylating and inhibiting its negative regulator PI3K23,31, and by mutations in tumour suppressor pro-
tuberous sclerosis 2 (TsC2; also known as tuberin)26. teins such as vHl32,33, succinate dehydrogenase (sDH)34
mTOr functions as a key metabolic integration point, and fumarate hydratase (FH)35. Once activated, HIF1
coupling growth signals to nutrient availability. Activated amplifies the transcription of genes encoding glucose
mTOr stimulates protein and lipid biosynthesis and cell transporters and most glycolytic enzymes, increasing
growth in response to sufficient nutrient and energy the capacity of the cell to carry out glycolysis36. In addi-
conditions and is often constitutively activated during tion, HIF1 activates the pyruvate dehydrogenase kinases
tumorigenesis29. At the molecular level, mTOr directly (PDKs), which inactivate the mitochondrial pyruvate
stimulates mrnA translation and ribosome biogenesis, dehydrogenase complex and thereby reduce the flow
and indirectly causes other metabolic changes by acti- of glucose-derived pyruvate into the tricarboxylic acid
vating transcription factors such as hypoxia-inducible (TCA) cycle37–39 (FIG. 2). This reduction in pyruvate flux
factor 1 (HIF1) even under normoxic conditions. into the TCA cycle decreases the rate of oxidative phos-
The subsequent HIF1-dependent metabolic changes phorylation and oxygen consumption, reinforcing the
are a major determinant of the glycolytic phenotype glycolytic phenotype and sparing oxygen under hypoxic
downstream of PI3K, AKT1 and mTOr (FIG. 2). conditions.
Inhibitors of HIF1 or the PDKs could potentially
HIF1 and MYC. The HIF1 and HIF2 complexes are the reverse some of the metabolic effects of tumorigenic HIF1
major transcription factors that are responsible for gene signalling and several such candidates, including the PDK
expression changes during the cellular response to low inhibitor dichloroacetic acid (DCA), are currently under
oxygen conditions. They are heterodimers that are com- evaluation for their therapeutic utility 40–43.
posed of the constitutively expressed HIF1β (also known In addition to its well-described role in controlling
as ArnT) subunit, and either the HIF1α or the HIF2α cell growth and proliferation, the oncogenic transcrip-
(also known as ePAs1) subunits, which are rapidly tion factor MyC also has several important effects on cell
stabilized on exposure to hypoxia30. under normoxic metabolism44. With respect to glycolysis, highly expressed
oncogenic MyC has been shown to collaborate with HIF
in the activation of several glucose transporters and
Genetic alterations Tumour microenvironment glycolytic enzymes, as well as lactate dehydrogenase A
(affecting p53, MYC, (hypoxia, pH, nutrients
AMPK, PI3K and HIF1) and autophagy) (lDHA) and PDK1 (ReFS 45,46). However, MyC also
activates the transcription of targets that increase mito-
Abnormal chondrial biogenesis and mitochondrial function, espe-
metabolic cially the metabolism of glutamine, which is discussed
phenotype in further detail below 47.

AMP-activated protein kinase. AMP-activated protein


Bioenergetics Biosynthesis Redox kinase (AMPK) is a crucial sensor of energy status and
has an important pleiotropic role in cellular responses
Figure 1 | Determinants of the tumour metabolic phenotype.Nature Reviews | Cancer
The metabolic to metabolic stress. The AMPK pathway couples energy
phenotype of tumour cells is controlled by intrinsic genetic mutations and external status to growth signals; biochemically, AMPK opposes
responses to the tumour microenvironment. Oncogenic signalling pathways controlling the effects of AKT1 and functions as a potent inhibitor
growth and survival are often activated by the loss of tumour suppressors (such as p53) or of mTOr (FIG. 2). The AMPK complex thus functions as
the activation of oncoproteins (such as PI3K). The resulting altered signalling modifies a metabolic checkpoint, regulating the cellular response
cellular metabolism to match the requirements of cell division. Abnormal
to energy availability. During periods of energetic stress,
microenvironmental conditions such as hypoxia, low pH and/or nutrient deprivation
elicit responses from tumour cells, including autophagy, which further affect metabolic AMPK becomes activated in response to an increased
activity. These adaptations optimize tumour cell metabolism for proliferation by AMP/ATP ratio, and is responsible for shifting cells to an
providing appropriate levels of energy in the form of ATP, biosynthetic capacity and the oxidative metabolic phenotype and inhibiting cell prolif-
maintenance of balanced redox status. AMPK, AMP-activated protein kinase; HIF1, eration48–50. Tumour cells must overcome this checkpoint
hypoxia-inducible factor 1. in order to proliferate in response to activated growth

nATure revIeWs | CanCer vOluMe 11 | FebruAry 2011 | 87

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

signalling pathways, even in a less than ideal microen- activated by oncogenes and the loss of tumour sup-
vironment 49. several oncogenic mutations and signal- pressors. Accordingly, many cancer cells exhibit a loss
ling pathways can suppress AMPK signalling 49, which of appropriate AMPK signalling: an event that may also
uncouples fuel signals from growth signals, allowing contribute to their glycolytic phenotype.
tumour cells to divide under abnormal nutrient condi- Given the role of AMPK, it is not surprising that
tions. This uncoupling permits tumour cells to respond STK11, which encodes liver kinase b1 (lKb1) — the
to inappropriate growth signalling pathways that are upstream kinase necessary for AMPK activation —
has been identified as a tumour suppressor gene and is
mutated in Peutz–Jeghers syndrome51. This syndrome
a Quiescent normal cell
is characterized by the development of benign gastro-
Lactate Glucose intestinal and oral lesions and an increased risk of
developing a broad range of malignancies. lKb1 is also
MCT GLUT
frequently mutated in sporadic cases of non-small-cell
Glucose lung cancer 52 and cervical carcinoma53. recent evidence
PI3K p53 LKB1
suggests that lKb1 mutations are tumorigenic owing to
Glycolysis PTEN the resulting decrease in AMPK signalling and loss of
AKT AMPK mTOr inhibition49. The loss of AMPK signalling allows
Lactate Pyruvate the activation of mTOr and HIF1, and therefore might
mTOR also support the shift towards glycolytic metabolism.
Clinically, there is currently considerable interest in eval-
TIGAR PDH PDK
uating whether AMPK agonists can be used to re-couple
p53 OCT1 MYC HIF fuel and growth signals in tumour cells and to shut down
Acetyl-CoA TCA
SCO2 cell growth. Two such agonists are the commonly used
antidiabetic drugs metformin and phenformin49,54–56. It
remains to be seen whether these agents represent a useful
b Proliferating tumour cell class of metabolic modifiers with antitumour activity.
Lactate Glucose
p53 and OCT1. Although the transcription factor and
GLUT tumour suppressor p53 is best known for its functions
MCT
in the DnA damage response (DDr) and apoptosis, it is
Glucose PI3K p53 LKB1 becoming clear that p53 is also an important regulator of
metabolism57. p53 activates the expression of hexokinase 2
PTEN
Glycolysis (HK2), which converts glucose to glucose-6-phosphate
AKT AMPK (G6P)58. G6P then either enters glycolysis to produce
PKM2
ATP, or enters the pentose phosphate pathway (PPP),
mTOR
Lactate Pyruvate which supports macromolecular biosynthesis by produc-
ing reducing potential in the form of reduced nicotina-
TIGAR PDH PDK mide adenine dinucleotide phosphate (nADPH) and/or
ribose, the building blocks for nucleotide synthesis.
p53 OCT1 MYC HIF
Acetyl-CoA TCA However, p53 inhibits the glycolytic pathway by upreg-
SCO2 ulating the expression of TP53-induced glycolysis and
apoptosis regulator (TIGAr), an enzyme that decreases
Figure 2 | Molecular mechanisms driving the Warburg effect. Relative to normal cells the levels of the glycolytic activator fructose-2,6-
Nature
(part a) the shift to aerobic glycolysis in tumour cells (part b) is driven Reviews | Cancer
by multiple bisphosphate59 (FIG. 2). Wild-type p53 also supports the
oncogenic signalling pathways. PI3K activates AKT, which stimulates glycolysis by directly expression of PTen, which inhibits the PI3K pathway,
regulating glycolytic enzymes and by activating mTOR. The liver kinase B1 (LKB1) tumour
thereby suppressing glycolysis (as discussed above)60.
suppressor, through AMP-activated protein kinase (AMPK) activation, opposes the
glycolytic phenotype by inhibiting mTOR. mTOR alters metabolism in a variety of ways, Furthermore, p53 promotes oxidative phosphorylation
but it has an effect on the glycolytic phenotype by enhancing hypoxia-inducible factor 1 by activating the expression of sCO2, which is required
(HIF1) activity, which engages a hypoxia-adaptive transcriptional programme. HIF1 for the assembly of the cytochrome c oxidase complex
increases the expression of glucose transporters (GLUT), glycolytic enzymes and pyruvate of the electron transport chain61. Thus, the loss of p53
dehydrogenase kinase, isozyme 1 (PDK1), which blocks the entry of pyruvate into the might also be a major force behind the acquisition of the
tricarboxylic acid (TCA) cycle. MYC cooperates with HIF in activating several genes that glycolytic phenotype.
encode glycolytic proteins, but also increases mitochondrial metabolism. The tumour OCT1 (also known as POu2F1) is a transcription
suppressor p53 opposes the glycolytic phenotype by suppressing glycolysis through factor, the expression of which is increased in several
TP53-induced glycolysis and apoptosis regulator (TIGAR), increasing mitochondrial human cancers, and it may cooperate with p53 in regu-
metabolism via SCO2 and supporting expression of PTEN. OCT1 (also known as POU2F1)
lating the balance between oxidative and glycolytic
acts in an opposing manner to activate the transcription of genes that drive glycolysis and
suppress oxidative phosphorylation. The switch to the pyruvate kinase M2 (PKM2) isoform metabolism62–64. The transcriptional programme that is
affects glycolysis by slowing the pyruvate kinase reaction and diverting substrates into initiated by OCT1 supports resistance to oxidative stress
alternative biosynthetic and reduced nicotinamide adenine dinucleotide phosphate and this may cooperate with the loss of p53 during trans-
(NADPH)-generating pathways. MCT, monocarboxylate transporter; PDH, pyruvate formation64. Data from studies of knockout mice and
dehydrogenase. The dashed lines indicate loss of p53 function. human cancer cell lines show that OCT1 regulates a set

88 | FebruAry 2011 | vOluMe 11 www.nature.com/reviews/cancer

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

of genes that increase glucose metabolism and reduce Glucose


mitochondrial respiration. One of these genes encodes
an isoform of PDK (PDK4) that has the same function
PPP
as the PDK enzymes that are activated by HIF1 (ReF. 64) NADPH
(FIG. 2). Although the mechanisms by which OCT1 is G6P
upregulated in tumour cells are poorly understood,
Glycolysis Macromolecules
its downstream effectors may be potential targets for
therapeutic intervention.
PEP

Beyond the Warburg effect ATP PKM2


Metabolic adaptation in tumours extends beyond the
Warburg effect. It is becoming clear that alterations to Pyruvate
metabolism balance the need of the cell for energy with
its equally important need for macromolecular building Figure 3 | PKM2 and its effect on glycolysis and the
pentose phosphate pathway. Pyruvate Naturekinase
Reviews | Cancer
isoform
blocks and maintenance of redox balance.
M2 (PKM2) is present in very few types of proliferating
normal cells but is present at high levels in cancer cells.
Pyruvate kinase (PK). As previously discussed, the gen- PKM2 catalyses the rate-limiting step of glycolysis,
eration of energy in the form of ATP through aerobic controlling the conversion of phosphoenolpyruvate (PEP)
glycolysis is required for unrestricted cancer cell pro- to pyruvate, and thus ATP generation. Although
liferation7. However, studies of the M2 isoform of PK counterintuitive, PKM2 opposes the Warburg effect by
(PKM2) have shown that ATP generation by aerobic inhibiting glycolysis and the generation of ATP in tumours.
glycolysis is not the sole metabolic requirement of a Although such an effect might at first seem to be
cancer cell, and that alterations to metabolism not only detrimental to tumour growth, the opposite is true. By
bolster ATP resources but also stimulate macromolecular slowing the passage of metabolites through glycolysis,
PKM2 promotes the shuttling of these substrates through
biosynthesis and redox control.
the pentose phosphate pathway (PPP) and other
PK catalyses the rate-limiting, ATP-generating step of alternative pathways so that large quantities of reduced
glycolysis in which phosphoenolpyruvate (PeP) is con- nicotinamide adenine dinucleotide phosphate (NADPH)
verted to pyruvate65. Multiple isoenzymes of PK exist in and other macromolecules are produced. These molecules
mammals: type l, which is found in the liver and kid- are required for macromolecule biosynthesis and the
neys; type r, which is expressed in erythrocytes; type maintenance of redox balance that is needed to support
M1, which is found in tissues such as muscle and brain; the rapid cell division that occurs within a tumour. G6P,
and type M2, which is present in self-renewing cells such glucose-6-phosphate.
as embryonic and adult stem cells65. Intriguingly, PKM2
is also expressed by many tumour cells. Furthermore,
it was discovered that although PKM1 could efficiently found to promote preferential expression of PKM2 over
promote glycolysis and rapid energy generation, PKM2 PKM1 by modulating exon splicing. The inclusion of
Pentose phosphate pathway is characteristically found in an inactive state and is exon 9 in the PK mrnA leads to translation of the PKM1
PPP. Biochemical pathway ineffective at promoting glycolysis66–68. isoform, whereas inclusion of exon 10 produces PKM2
converting glucose into This observation was ignored by the scientific com- (ReF. 71). MyC upregulates the expression of heteroge-
substrates for nucleotide munity for several years owing to its shear counterin- neous nuclear ribonucleoproteins (hnrnPs) that bind
biosynthesis and redox control,
such as ribose and NADPH.
tuitive nature: a tumour-specific glycolytic enzyme that to exon 9 of the PK mrnA and lead to the preferen-
Owing to multiple connections inhibits ATP generation and antagonizes the Warburg tial inclusion of exon 10 and thus to the predominant
to the glycolytic pathway, the effect. Only on closer examination of the full metabolic production of PKM2. by promoting PKM2 expression,
PPP can operate in various requirements of a cancer cell was the advantage of PKM2 MyC promotes the production of nADPH in order to
modes to allow the production
expression revealed. A cancer cell, like any normal cell, match the increased ATP production and to satisfy the
of NADPH and/or ribose as
required. must obtain the building blocks that are required for the auxiliary needs required for increased proliferation.
synthesis of lipids, nucleotides and amino acids. Without At the clinical level, increased PKM2 expression has
Macromolecular sufficient precursors available for this purpose, rapid cell been documented in patient samples of various cancer
biosynthesis proliferation will halt, no matter how vast a supply of types, leading to the proposal that PKM2 might be a use-
Biochemical synthesis of the
carbohydrates, nucleotides,
ATP is present. PKM2 provides an advantage to cancer ful biomarker for the early detection of tumours65,72–74.
proteins and lipids that make cells because, by slowing glycolysis, this isozyme allows However, further study of the prevalence of PKM2 in
up cells and tissues. These carbohydrate metabolites to enter other subsidiary cancers and the effect of PKM2 on tumorigenesis is still
pathways require energy, pathways, including the hexosamine pathway, uridine required.
reducing power and
diphosphate (uDP)–glucose synthesis, glycerol syn-
appropriate substrates.
thesis and the PPP, which generate macromolecule NADPH. A key molecule produced as a result of the
Reduced nicotinamide precursors, that are necessary to support cell prolif- promotion of the oxidative PPP by PKM2 is nADPH
adenine dinucleotide eration, and reducing equivalents such as nADPH4,28,69 (FIG. 4). nADPH functions as a cofactor and provides
phosphate (FIG. 3). subsequent studies have confirmed that PKM2 reducing power in many enzymatic reactions that are
NADPH. Cofactor that drives
anabolic biochemical reactions
expression by lung cancer cells confers a tumorigenic crucial for macromolecular biosynthesis. Although
and provides reducing capacity advantage over cells expressing the PKM1 isoform70. other metabolites are produced as a result of increased
to combat oxidative stress. Interestingly, the classical oncoprotein MyC has been PPP activity, including ribose, which can be converted

nATure revIeWs | CanCer vOluMe 11 | FebruAry 2011 | 89

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Glucose Isocitrate Malate Glutamine


Isocitrate dehydrogenases. Another mechanism by
which nADPH is produced in mammalian cells is the
IDH1 or reaction converting isocitrate to α-ketoglutarate (αKG),
ME1
IDH2 which is catalysed by nADP-dependent isocitrate dehy-
drogenase 1 (IDH1) and IDH2. IDH1 and IDH2 are
G6P αKG Pyruvate Glutamate homodimeric enzymes that act in the cytoplasm and
mitochondria, respectively, to produce nADPH by this
PPP reaction. IDH1 and IDH2 are highly homologous and
MYC Glutaminolysis
PKM2 structurally and functionally distinct from the nAD-
dependent enzyme IDH3, which functions in the TCA
cycle to produce the nADH that is required for oxidative
MYC NADPH GSH phosphorylation.
It has recently been found that specific mutations
in IDH1 and IDH2 are linked to tumorigenesis. Two
Redox control
independent cancer genome sequencing projects iden-
tified driver mutations in IDH1 in glioblastoma and
Figure 4 | Mechanisms of redox control and their alterationsNaturein cancer. The| Cancer
Reviews acute myeloid leukaemia (AMl)3,77. subsequent stud-
production of two of the most abundant antioxidants, reduced nicotinamide adenine ies revealed that IDH1 or IDH2 is mutated in approxi-
dinucleotide phosphate (NADPH) and glutathione (GSH), has been shown to be mately 80% of adult grade II and grade III gliomas and
modulated in cancers. Pyruvate kinase isoform M2 (PKM2), which is overexpressed in secondary glioblastomas, and in approximately 30% of
many cancer cells, can divert metabolic precursors away from glycolysis and into the
cytogenetically normal cases of AMl78–80. The IDH1 and
pentose phosphate pathway (PPP) to produce NADPH. NADP-dependent isocitrate
dehydrogenase 1 (IDH1), IDH2 and malic enzyme 1 (ME1) also contribute to NADPH
IDH2 mutations associated with the development of
production. MYC increases glutamine uptake and glutaminolysis, driving the de novo glioma and AMl are restricted to crucial arginine resi-
synthesis of GSH. Additionally, MYC contributes to NADPH production by promoting the dues required for isocitrate binding in the active site of
expression of PKM2. Together, NADPH and GSH control increased levels of reactive the protein: r132 in IDH1, and r172 and r140 in IDH2
oxygen species (ROS) driven by increased cancer cell proliferation. αKG, α-ketoglutarate; (ReFS 3,77,79,80). Affected patients are heterozygous for
G6P, glucose-6-phosphate. these mutations, suggesting that these alterations may
cause an oncogenic gain-of-function. The range of muta-
tions differs in the two diseases, with the IDH1 r132H
into nucleotides, the supply of these building blocks mutation predominating in gliomas (>90%), whereas a
may not be as important as the production of nADPH. more diverse collection of mutations in both IDH1 and
not only does nADPH fuel macromolecular biosyn- IDH2 are found in AMl4,78–80.
thesis, but it is also a crucial antioxidant, quenching the It was initially proposed that these mutations might
reactive oxygen species (rOs) produced during rapid act through dominant-negative inhibition of IDH1
cell proliferation. In particular, nADPH provides the and IDH2 activity, which could lead to a reduction in
reducing power for both the glutathione (GsH) and cytoplasmic αKG concentration, inhibition of prolyl
thioredoxin (TrX) systems that scavenge rOs hydroxylase activity and stabilization of HIF1 (ReF. 81).
and repair rOs-induced damage75. The double-pronged However, it has recently been shown that these muta-
importance of nADPH in cancer cell metabolism has tions cause IDH1 and IDH2 to acquire a novel enzy-
prompted proposals of clinical intervention by inhibit- matic activity that converts αKG to 2-hydroxyglutarate
ing nADPH production. Attenuation of the PPP would (2-HG) in a nADPH-dependent manner 79,80,82 (FIG. 5).
theoretically dampen nADPH production in cancer cells, In fact, this change causes the mutated IDH1 and IDH2
slowing macromolecular biosynthesis and rendering the enzymes to switch from nADPH production to nADPH
transformed cells vulnerable to free radical-mediated consumption, with potentially important consequences
damage. In this way, the advantage conferred by PKM2 for the cellular redox balance. The product of the novel
expression would be eliminated. In preclinical studies, reaction, 2-HG, is a poorly understood metabolite. 2-HG
drugs such as 6-amino-nicotinamide (6-An), which is present at low concentrations in normal cells and
inhibits G6P dehydrogenase (G6PD; the enzyme that tissues. However, in patients with somatic IDH1 or IDH2
initiates the PPP) have demonstrated anti-tumorigenic mutations, 2-HG builds up to high levels in glioma tis-
effects in leukaemia, glioblastoma and lung cancer cell sues, and in the leukaemic cells and sera of patients with
lines76. However, additional basic research and complete AMl79,80,82. It remains to be determined whether these
clinical trials will be required to properly assess their high concentrations of 2-HG are mechanistically respon-
therapeutic potential. sible for the ability of IDH1 and IDH2 mutations to drive
The discovery and subsequent investigation of the tumorigenesis. Importantly, levels of αKG, isocitrate and
2‑hydroxyglutarate effects of PKM2 expression has shown that we must several other TCA metabolites are not altered in cell
2-HG. A dicarboxylic acid construct a post-Warburg model of cancer metabo- lines or tissues expressing IDH1 mutations, suggesting
metabolite produced from αKG lism, in which ATP generation is not the sole metabolic that other metabolic pathways can adjust and maintain
by the NADPH-dependent requirement of tumour cells. This turning point has led normal levels of these essential metabolites79,82.
reaction of the mutated forms
of IDH1 and IDH2. It is also
to the realization that the metabolic alterations present studies of IDH1 and IDH2 have established a new
produced at low levels by other in cancer cells promote not only ATP resources, but also paradigm in oncogenesis: a driver mutation that con-
enzymes. macromolecular biosynthesis and redox control (FIG. 1). fers a new metabolic enzymatic activity that produces a

90 | FebruAry 2011 | vOluMe 11 www.nature.com/reviews/cancer

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

potential oncometabolite. The molecular mechanisms by proliferative cancer cells, rOs regulation is crucial owing
which IDH1 and IDH2 mutations contribute to tumori- to the presence of oncogenic mutations that promote
genesis are still under investigation, as is the possibil- aberrant metabolism and protein translation, result-
ity that these mutant enzymes may be useful targets for ing in increased rates of rOs production. Transformed
therapy. Curiously, although IDH1 and IDH2 mutations cells counteract this accumulation of rOs by further
are clearly powerful drivers of glioma and AMl, they upregulating antioxidant systems, seemingly creating a
seem to be rare or absent in other tumour types78,83,84. paradox of high rOs production in the presence of high
This observation highlights the importance of the antioxidant levels95–98 (FIG. 6).
specific cellular context in understanding metabolic
perturbations in cancer cells. RB, PTEN and p53. There is currently a scientific con-
sensus that cancer cells alter their metabolic pathways
Metabolic alterations supporting redox status and regulatory mechanisms so that rOs and antioxi-
rOs are a diverse class of radical species that are pro- dants are tightly controlled and maintained at higher
duced in all cells as a normal byproduct of metabolic levels than in normal cells. However, during the process
processes. rOs are heterogeneous in their properties of tumorigenesis, loss of tumour suppressors may cause
and have a plethora of downstream effects, depending cells to become overloaded with the products of aberrant
on the concentrations at which they are present. metabolism and lose control of redox balance. For exam-
At low levels, rOs increase cell proliferation and ple, when the tumour suppressor TSC2 is deleted, mTOr
survival through the post-translational modification of becomes hyperactivated99. Hyperactivated mTOr leads
kinases and phosphatases85–87. The production of this to an upregulation of translation and increased rOs
low level of rOs can be driven by nADPH and nADPH production100. In a cancer cell that has additionally lost
oxidase (nOX) and is required for homeostatic signal- function of the tumour suppressor retinoblastoma (rb),
ling events. At moderate levels, rOs induce the expres- which normally participates in the antioxidant response,
sion of stress-responsive genes such as HIF1Α, which in the increased rOs production is not countered and the
turn trigger the expression of proteins providing pro- cell will undergo apoptosis99. similar results have been
survival signals, such as the glucose transporter GluT1 seen with loss of PTen, and hyperactivation of AKT1
(also known as slC2A1) and vascular endothelial leads to FOXO inactivation and increased oxidative
growth factor (veGF)88,89. However, at high levels, rOs stress101.
can cause damage to macromolecules, including DnA; A comparable theory can be proposed for p53. p53
induce the activation of protein kinase Cδ (PKCδ), trig- may promote oxidative stress while inducing apopto-
gering senescence90,91; and/or cause permeabilization of sis102–104, but it also has an important role in reducing oxi-
the mitochondria, leading to the release of cytochrome c dative stress as a defence mechanism105,106. Glutaminase 2
and apoptosis92,93. Cells counteract the detrimental (Gls2) is upregulated by p53 and drives de novo synthesis
effects of rOs by producing antioxidant molecules, of GsH107. Furthermore, through the p53 target gene
such as reduced GsH and TrX. These molecules reduce cyclin-dependent kinase inhibitor 1A (CDKN1A, which
excessive levels of rOs to prevent irreversible cellular encodes p21), p53 promotes the stabilization of the trans-
damage94. Importantly, several of these antioxidant sys- cription factor nrF2 (also known as nFe2l2)108. nrF2
tems, including GsH and TrX, rely on the reducing is the master antioxidant transcription factor and upreg-
power of nADPH to maintain their activities. In highly ulates the expression of several antioxidant and detoxify-
ing molecules108. When rOs levels are low, nrF2 binds
to kelch-like eCH-associated protein 1 (KeAP1), which
Glucose Acetyl-CoA Fatty acids triggers nrF2 degradation. under oxidative stress, p53 is
activated and stimulates expression of p21. p21 prevents
the KeAP1–nrF2 interaction and preserves nrF2,
Lactate Pyruvate Citrate Isocitrate
driving antioxidant countermeasures108. loss of p53 in
NADP a cancer cell inactivates this redox maintenance mecha-
IDH1 or
Acetyl-CoA IDH2 nism: because p21 is not activated, nrF2 continues to be
TCA NADPH NADP degraded, antioxidant proteins are not expressed and the
αKG 2-HG redox balance is lost. From a clinical point of view, it may
IDH1 mutant or
be possible to exploit loss-of-function p53 mutations or
Glutamine or glutamate IDH2 mutant other tumour suppressor genes by applying additional
oxidative stress. In the absence of the redox maintenance
Figure 5 | IDH1 and IDH2 mutations cause an oncometabolic gain of function. pathway that is supported by these tumour suppressors,
Nature
Certain somatic mutations at crucial arginine residues in isocitrate Reviews | Cancer
dehydrogenase 1 malignant cells might be selectively killed109–111.
(IDH1, which is cytoplasmic) and IDH2 (which is mitochondrial) are common early driver
mutations in glioma and acute myeloid leukaemia (AML). These mutations are unusual
DJ1. Much of the research involving rOs and oxidative
because they cause the gain of a novel enzymatic activity. Instead of isocitrate being
converted to α-ketoglutarate (αKG) with the production of reduced nicotinamide stress has emerged from work in the field of neurode-
adenine dinucleotide phosphate (NADPH), αKG is converted to 2-hydroxyglutarate generative diseases. Only recently has it been realized
(2-HG) with the consumption of NADPH. 2-HG builds up to high levels in tumour cells that similar mechanisms maintain appropriate redox
and tissues of affected patients and supports tumour progression by a mechanism that is status in both normal neurons and cancer cells. One
yet to be determined. TCA, tricarboxylic acid cycle. protein involved in preventing neurodegeneration that

nATure revIeWs | CanCer vOluMe 11 | FebruAry 2011 | 91

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

• Proliferation • Adaptive genes • Senescence cancers. Additional investigation of the cancer risk of
• Cell survival • Mutagenesis • Cell death patients with other neurodegenerative disorders, such as
Als, may provide key insights into potential therapeutic

ROS level
exploitation of the heightened need to maintain redox
balance in a cancer cell.
Cancer cell
Glutamine and MYC. It has long been known that cell
• Metabolism culture medium must be supplemented with high con-
• Protein translation Antioxidants
centrations of glutamine to support robust cell prolif-
eration120–122. However, it has recently been shown that
Figure 6 | relationship between the levels of rOS and cancer. Nature
TheReviews
effect of| Cancer transformation stimulates glutaminolysis and that many
reactive oxygen species (ROS) on cell fate depends on the level at which ROS are tumour cells are critically dependent on this amino
present. Low levels of ROS (yellow) provide a beneficial effect, supporting cell acid123,124. After glutamine enters the cell, glutaminase
proliferation and survival pathways. However, once levels of ROS become excessively enzymes convert it to glutamate, which has several fates.
high (purple), they cause detrimental oxidative stress that can lead to cell death. To Glutamate can be converted directly into GsH by the
counter such oxidative stress, a cell uses antioxidants that prevent ROS from enzyme glutathione cysteine ligase (GCl) (FIG. 4). reduced
accumulating at high levels. In a cancer cell, aberrant metabolism and protein
GsH is one of the most abundant antioxidants found in
translation generate abnormally high levels of ROS. Through additional mutations and
adaptations, a cancer cell exerts tight regulation of ROS and antioxidants in such a way mammalian cells and is vital to controlling the redox state
that the cell survives and the levels of ROS are reduced to moderate levels (blue). This of all subcellular compartments97. Glutamate can also be
extraordinary control of ROS and the mechanisms designed to counter it allow the converted to αKG and enter the TCA cycle. This pro-
cancer cell to avoid the detrimental effects of high levels of ROS, but also increase the cess of anapleurosis supplies the carbon input required
chance that the cell will experience additional ROS-mediated mutagenic events and for the TCA cycle to function as a biosynthetic ‘hub’ and
stress responses that promote tumorigenesis. Figure inspired by discussions with permits the production of other amino acids and fatty
Navdeep Chandel, Northwestern University, Chicago, USA. acids. There is also recent evidence that some glutamine-
derived carbon can exit the TCA cycle as malate and
serve as a substrate for malic enzyme 1 (Me1), which
has also been investigated in the context of cancer is DJ1 produces nADPH125. The precise mechanisms regulating
(also known as PArK7). similar to p21, DJ1 stabilizes the fate of glutamine in tumour cells are not completely
nrF2 and thereby promotes antioxidant responses112. understood, and it is likely that genetic background and
DJ1 is mutated and inactive in several neurodegenera- microenvironmental factors have a role.
tive disorders, most notably Parkinson’s disease113. In One factor that is known to have a major role in regu-
these disorders, it is believed that loss of DJ1 func- lating glutaminolysis is MyC, further supporting the con-
tion leads to elevated oxidative stress in the brain and cept that MyC promotes not only proliferation but also
increased neuronal cell death114. In the context of cancer, the production of accompanying macromolecules and
PARK7 has been described as an oncogene115. In patients antioxidants that are required for growth. MyC increases
Parkinson’s disease
with lung, ovarian and oesophageal cancers, high DJ1 glutamine uptake by directly inducing the expression of the
A neurodegenerative disorder
affecting the CNS, which is expression in the tumour predicts a poor outcome115–117. glutamine transporters slC5A1 and slC7A1 (also known
characterized by muscle At a mechanistic level, DJ1 stimulates AKT1 activity as CAT1)124. Furthermore, MyC indirectly increases the
rigidity and the onset of both in vitro and in vivo by regulating the function of level of glutaminase 1 (Gls1), the first enzyme of glutami-
tremors. the tumour suppressor PTen115. Although this func- nolysis, by repressing the expression of microRNA‑23A
Amyotrophic lateral
tion seems to be a logical candidate for the mechanism and microRNA‑23B, which inhibit GLS1 (ReF. 124). Thus,
sclerosis underlying the tumorigenic role of DJ1, high DJ1 expres- MyC may support antioxidant capacity by driving PPP-
ALS. Also known as Lou sion may also promote tumorigenesis by reducing the based nADPH production through promoting the expres-
Gehrig’s disease; it occurs oxidative stress caused by aberrant cell proliferation and sion of the PKM2 isoform, as described above, and also by
owing to the degeneration of
thereby prevent rOs-induced cell death. increasing the synthesis of GsH through glutaminolysis
the CNS and leads to the
inability to control muscles and several other proteins that are inactivated in neuro- (FIG. 4). A comprehensive and quantitative investigation
eventual muscle atrophy. degenerative disorders have antioxidant properties, of glutamine metabolism in patient samples has not yet
including the enzyme superoxide dismutase 1 (sOD1). been reported. However, new techniques for measuring
Glutaminolysis Mutations in sOD1 are responsible for 20% of familial glutamine and its metabolites have been developed and
The catabolic metabolism of
glutamine, which yields
cases of amyotrophic lateral sclerosis (Als)118. However, it should soon permit the detailed examination of glutamine
substrates that replenish the is still unknown whether sOD1 or other key antioxidant metabolism and MyC expression in patient tumours126.
TCA cycle, produce GSH and enzymes are hyperactivated in cancer cells and whether Furthermore, work is underway to determine whether
supply building blocks for they have important roles in tumorigenesis. supporting other oncoproteins such as PI3K and srC have a role in
amino acid and nucleotide
the notion that loss of DJ1 prevents appropriate redox promoting glutaminolysis. supporting this theory, it has
synthesis.
control in cancers, an inverse correlation has been been shown that cells with a hyperactive ras oncogene
Anapleurosis reported between cancer risk and Parkinson’s disease. A require a stable flow of glutamine and GsH generation in
Category of reactions that recent meta-analysis of patients with Parkinson’s disease order to balance redox demands13,111. It is also interesting
serve to replenish the determined that they have an approximately 30% lower to speculate that part of the mechanism responsible for the
intermediate substrates of an
anabolic biochemical pathway,
risk of developing cancers compared with controls119. clinical efficacy of l-asparaginase in treating certain leu-
especially important in the TCA This lower risk was associated with several different kaemias may be related to this phenomenon, as l-aspara-
cycle. cancer types, including lung, prostate and colorectal ginase therapy reduces serum levels of both asparagine and

92 | FebruAry 2011 | vOluMe 11 www.nature.com/reviews/cancer

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

glutamine127,128. nevertheless, several questions regarding biochemical conditions in the tumour influence
the role of glutamine in tumorigenesis remain to be cellular metabolism. because metabolite concentra-
answered. tions are governed by both supply by the vasculature
and demand by the tissue, changes in metabolism of
Metabolic adaptation to the microenvironment both the tumour and normal stromal cells also have
In addition to the genetic changes that alter tumour cell a profound effect on microenvironmental condi-
metabolism, the abnormal tumour microenvironment has tions (FIG. 1). The complex and dynamic relationship
a major role in determining the metabolic phenotype of between tumour metabolism and the microenviron-
tumour cells. Tumour vasculature is structurally and func- ment emphasizes the importance of studying metabolic
tionally abnormal, and combined with intrinsically altered regulation in vivo using appropriate model systems, as
tumour cell metabolism, creates spatial and temporal het- well as the need for more sophisticated measurements
erogeneity in oxygenation, pH, and the concentrations of of cell metabolism and relevant microenvironmental
glucose and many other metabolites. These extreme con- conditions in human tumours.
ditions induce a collection of cellular stress responses that
further contribute to the distorted metabolic phenotype of Metabolic flexibility. Although aerobic glycolysis (the
tumour cells and influence tumour progression129. Warburg effect) is the best documented metabolic phe-
notype of tumour cells, it is not a universal feature of all
Response to hypoxia. The response to hypoxia is the best human cancers140. Moreover, even in glycolytic tumours,
studied of tumour cell stress responses owing to the well- oxidative phosphorylation is not completely shut down.
known effects of hypoxia on tumour radioresistance and It is clear from both clinical FDG–PeT data, as well as
metastasis. Consequently, tumour hypoxia is a poor prog- in vitro and in vivo experimental studies, that tumour
nostic factor in a number of malignancies6,129–131. several cells are capable of using alternative fuel sources. In fact,
molecular pathways that influence cellular metabolism up to 30% of tumours are considered FDG–PeT-negative
are altered under hypoxia. As described above, hypoxia depending on the tumour type16,17. Amino acids, fatty
alters transcription through the stabilization of HIF, acids and even lactate have been shown to function as
which increases glycolytic capacity and decreases mito- fuels for tumour cells in certain genetic and microen-
chondrial respiration132. In addition, and independently vironmental contexts125,141,142. The carnitine palmitoyl-
of HIF, hypoxia inhibits signalling through mTOr, which transferase enzymes that regulate the β-oxidation of
is a major regulator of multiple mechanisms contribut- fatty acids may have a key role in determining some
ing to the altered metabolic phenotype133,134. specifically, of these phenotypes. Furthermore, owing to the dynamic
the induction of autophagy may be of crucial impor- nature of the tumour microenvironment, it is likely that
tance135. Although mTOr inhibition would usually be the metabolic phenotype of tumour cells changes to
considered tumour suppressive, there is evidence that adapt to the prevailing local conditions. The regulation
in advanced malignancies such a response can increase of this metabolic flexibility is poorly understood and will
the tolerance to hypoxia and promote tumour cell sur- require a much greater degree of understanding if effec-
vival during metabolic stress. This finding supports the tive therapeutic strategies targeting metabolism are to be
concept that, in certain microenvironmental or genetic developed and effectively deployed.
contexts, as in the case of rb inactivation, tumour cells
may benefit from retaining the ability to moderate Conclusion
mTOr signalling 99. Finally, extreme hypoxia (<0.02% O2) Mutations in oncogenes and tumour suppressor genes
causes endoplasmic reticulum stress and activates the cause alterations to multiple intracellular signalling
unfolded protein response, which provides a further pathways that affect tumour cell metabolism and re-
adaptive mechanism that allows tumour cells to survive engineer it to allow enhanced survival and growth. In
under adverse metabolic conditions134,136–138. fact, it is likely that metabolic alterations are required
Other metabolic stress conditions such as low pH for tumour cells to be able to respond to the prolifera-
and low glucose are also prevalent in solid tumours and tive signals that are delivered by oncogenic signalling
are likely to be major determinants of the metabolic pathways. In addition, the unique biochemical microen-
phenotype. The molecular pathways that are involved vironment further influences the metabolic phenotype
in responding to these conditions are currently under of tumour cells, and thus affects tumour progres-
investigation, which will undoubtedly enhance our sion, response to therapy and patient outcome. These
knowledge of the mechanistic determinants of tumour metabolic adaptations must balance the three crucial
cell metabolism. since it has been well established that requirements of tumour cells: increased energy produc-
microenvironmental factors affect sensitivity to radia- tion, sufficient macromolecular biosynthesis and main-
tion, traditional chemotherapy and targeted therapies, tenance of redox balance. Only by thoroughly dissecting
a better understanding of the diverse avenues of meta- these processes will we discover the Achilles heels of
bolic regulation in cancer cells may offer new oppor- tumour metabolic pathways and be able to translate this
tunities to modify the tumour microenvironment for knowledge to the development and implementation of
therapeutic gain139. novel classes of therapeutics. The ultimate goal is to
It should be noted that the relationship between the design treatment strategies that slow tumour progres-
tumour microenvironment and cancer cell metabo- sion, improve the response to therapy and result in a
lism is not one of simple cause and effect, in which positive clinical outcome.

nATure revIeWs | CanCer vOluMe 11 | FebruAry 2011 | 93

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

1. Stratton, M. R., Campbell, P. J. & Futreal, P. A. The 27. Khatri, S., Yepiskoposyan, H., Gallo, C. A., Tandon, P. 51. Jenne, D. E. et al. Peutz‑Jeghers syndrome is caused
cancer genome. Nature 458, 719–724 (2009). & Plas, D. R. FOXO3a regulates glycolysis via by mutations in a novel serine threonine kinase.
2. The International Cancer Genome Consortium. transcriptional control of tumor suppressor TSC1. Nature Genet. 18, 38–43 (1998).
International network of cancer genome projects. J. Biol. Chem. 285, 15960–15965 (2010). 52. Ji, H. et al. LKB1 modulates lung cancer
Nature 464, 993–998 (2010). 28. Fang, M. et al. The ER UDPase ENTPD5 promotes differentiation and metastasis. Nature 448, 807–810
3. Parsons, D. W. et al. An integrated genomic analysis of protein N‑glycosylation, the Warburg effect, and (2007).
human glioblastoma multiforme. Science 321, proliferation in the PTEN pathway. Cell 143, 711–724 53. Wingo, S. N. et al. Somatic LKB1 mutations promote
1807–1812 (2008). (2010). cervical cancer progression. PLoS ONE 4, e5137
Sequencing of the glioblastoma genome in which 29. Guertin, D. A. & Sabatini, D. M. Defining the role of (2009).
mutation of IDH1 was identified as a driver mTOR in cancer. Cancer Cell 12, 9–22 (2007). 54. Wang, W. & Guan, K. L. AMP‑activated protein kinase
mutation. 30. Bertout, J. A., Patel, S. A. & Simon, M. C. The impact and cancer. Acta Physiol. 196, 55–63 (2009).
4. Vander Heiden, M. G., Cantley, L. C. & Thompson, of O2 availability on human cancer. Nature Rev. 55. Libby, G. et al. New users of metformin are at low risk
C. B. Understanding the Warburg effect: the metabolic Cancer 8, 967–975 (2008). of incident cancer: a cohort study among people with
requirements of cell proliferation. Science 324, 31. Inoki, K., Corradetti, M. N. & Guan, K. L. type 2 diabetes. Diabetes Care 32, 1620–1625
1029–1033 (2009). Dysregulation of the TSC‑mTOR pathway in human (2009).
Provocative review advancing the concept that disease. Nature Genet. 37, 19–24 (2005). 56. Anisimov, V. N. et al. Effect of metformin on life span
glycolytic metabolism supports biosynthetic 32. Kapitsinou, P. P. & Haase, V. H. The VHL tumor and on the development of spontaneous mammary
pathways. suppressor and HIF: insights from genetic studies in tumors in HER‑2/neu transgenic mice. Exp. Gerontol.
5. Newsholme, E. A., Crabtree, B. & Ardawi, M. S. The mice. Cell Death Differ. 15, 650–659 (2008). 40, 685–693 (2005).
role of high rates of glycolysis and glutamine utilization 33. Kaelin, W. G. The von Hippel‑Lindau tumour 57. Vousden, K. H. & Ryan, K. M. p53 and metabolism.
in rapidly dividing cells. Biosci. Rep. 5, 393–400 suppressor protein: O2 sensing and cancer. Nature Nature Rev. Cancer 9, 691–700 (2009).
(1985). Rev. Cancer 8, 865–873 (2008). 58. Mathupala, S. P., Heese, C. & Pedersen, P. L. Glucose
6. Tatum, J. L. et al. Hypoxia: importance in tumor 34. Selak, M. A. et al. Succinate links TCA cycle catabolism in cancer cells. The type II hexokinase
biology, noninvasive measurement by imaging, and dysfunction to oncogenesis by inhibiting HIF‑α prolyl promoter contains functionally active response
value of its measurement in the management of hydroxylase. Cancer Cell 7, 77–85 (2005). elements for the tumor suppressor p53. J. Biol. Chem.
cancer therapy. Int. J. Radiat. Biol. 82, 699–757 35. King, A., Selak, M. A. & Gottlieb, E. Succinate 272, 22776–22780 (1997).
(2006). dehydrogenase and fumarate hydratase: linking 59. Bensaad, K. et al. TIGAR, a p53‑inducible regulator of
7. Warburg, O. On the origin of cancer cells. Science mitochondrial dysfunction and cancer. Oncogene 25, glycolysis and apoptosis. Cell 126, 107–120 (2006).
123, 309–314 (1956). 4675–4682 (2006). 60. Stambolic, V. et al. Regulation of PTEN transcription
8. Semenza, G. L. et al. ‘The metabolism of tumours’: 36. Semenza, G. L. HIF‑1: upstream and downstream of by p53. Mol. Cell 8, 317–325 (2001).
70 years later. Novartis Found. Symp. 240, 251–260; cancer metabolism. Curr. Opin. Genet. Dev. 20, 61. Matoba, S. et al. p53 regulates mitochondrial
discussion 260–254 (2001). 51–56 (2010). respiration. Science 312, 1650–1653 (2006).
9. Frezza, C. & Gottlieb, E. Mitochondria in cancer: not 37. Papandreou, I., Cairns, R. A., Fontana, L., Lim, A. L. & 62. Almeida, R. et al. OCT‑1 is over‑expressed in intestinal
just innocent bystanders. Semin. Cancer Biol. 19, Denko, N. C. HIF‑1 mediates adaptation to hypoxia by metaplasia and intestinal gastric carcinomas and
4–11 (2009). actively downregulating mitochondrial oxygen binds to, but does not transactivate, CDX2 in gastric
10. Weinhouse, S. The Warburg hypothesis fifty years consumption. Cell Metab. 3, 187–197 (2006). cells. J. Pathol. 207, 396–401 (2005).
later. Z. Krebsforsch. Klin. Onkol. Cancer Res. Clin. 38. Kim, J. W., Tchernyshyov, I., Semenza, G. L. & 63. Jin, T. et al. Examination of POU homeobox gene
Oncol. 87, 115–126 (1976). Dang, C. V. HIF‑1‑mediated expression of pyruvate expression in human breast cancer cells. Int. J. Cancer
11. Funes, J. M. et al. Transformation of human dehydrogenase kinase: a metabolic switch required for 81, 104–112 (1999).
mesenchymal stem cells increases their dependency cellular adaptation to hypoxia. Cell Metab. 3, 64. Shakya, A. et al. Oct1 loss of function induces a
on oxidative phosphorylation for energy production. 177–185 (2006). coordinate metabolic shift that opposes
Proc. Natl Acad. Sci. USA 104, 6223–6228 (2007). References 37 and 38 showed that HIF1 induces tumorigenicity. Nature Cell Biol. 11, 320–327 (2009).
12. Fogal, V. et al. Mitochondrial p32 protein is a critical expression of PDK1, which limits the flow of 65. Mazurek, S., Boschek, C. B., Hugo, F. & Eigenbrodt, E.
regulator of tumor metabolism via maintenance of pyruvate into the TCA cycle and decreases oxidative Pyruvate kinase type M2 and its role in tumor growth
oxidative phosphorylation. Mol. Cell. Biol. 30, phosphorylation. and spreading. Semin. Cancer Biol. 15, 300–308
1303–1318 (2010). 39. Lu, C. W., Lin, S. C., Chen, K. F., Lai, Y. Y. & Tsai, S. J. (2005).
13. Weinberg, F. et al. Mitochondrial metabolism and ROS Induction of pyruvate dehydrogenase kinase‑3 by 66. Mazurek, S., Zwerschke, W., Jansen‑Durr, P. &
generation are essential for Kras‑mediated hypoxia‑inducible factor‑1 promotes metabolic switch Eigenbrodt, E. Metabolic cooperation between
tumorigenicity. Proc. Natl Acad. Sci. USA 107, and drug resistance. J. Biol. Chem. 283, different oncogenes during cell transformation:
8788–8793 (2010). 28106–28114 (2008). interaction between activated ras and HPV‑16 E7.
14. Gatenby, R. A. & Gillies, R. J. Why do cancers have 40. Cairns, R. A. et al. Pharmacologically increased tumor Oncogene 20, 6891–6898 (2001).
high aerobic glycolysis? Nature Rev. Cancer 4, hypoxia can be measured by 18F‑Fluoroazomycin 67. Zwerschke, W. et al. Modulation of type M2 pyruvate
891–899 (2004). arabinoside positron emission tomography and kinase activity by the human papillomavirus type 16
15. Gillies, R. J., Robey, I. & Gatenby, R. A. Causes and enhances tumor response to hypoxic cytotoxin E7 oncoprotein. Proc. Natl Acad. Sci. USA 96,
consequences of increased glucose metabolism of PR‑104. Clin. Cancer Res. 15, 7170–7174 (2009). 1291–1296 (1999).
cancers. J. Nucl. Med. 49 (Suppl. 2), 24S‑42S (2008). 41. Michelakis, E. D., Webster, L. & Mackey, J. R. 68. Christofk, H. R., Vander Heiden, M. G., Wu, N.,
16. Gambhir, S. S. Molecular imaging of cancer with Dichloroacetate (DCA) as a potential metabolic‑ Asara, J. M. & Cantley, L. C. Pyruvate kinase M2 is a
positron emission tomography. Nature Rev. Cancer 2, targeting therapy for cancer. Br. J. Cancer 99, phosphotyrosine‑binding protein. Nature 452,
683–693 (2002). 989–994 (2008). 181–186 (2008).
17. Gambhir, S. S. et al. A tabulated summary of the FDG 42. Semenza, G. L. Defining the role of hypoxia‑inducible 69. Marshall, S., Bacote, V. & Traxinger, R. R. Discovery of
PET literature. J. Nucl. Med. 42, 1S–93S (2001). factor 1 in cancer biology and therapeutics. Oncogene a metabolic pathway mediating glucose‑induced
18. Jadvar, H., Alavi, A. & Gambhir, S. S. 18F‑FDG uptake 29, 625–634 (2010). desensitization of the glucose transport system. Role
in lung, breast, and colon cancers: molecular biology 43. Onnis, B., Rapisarda, A. & Melillo, G. Development of of hexosamine biosynthesis in the induction of insulin
correlates and disease characterization. J. Nucl. Med. HIF‑1 inhibitors for cancer therapy. J. Cell. Mol. Med. resistance. J. Biol. Chem. 266, 4706–4712 (1991).
50, 1820–1827 (2009). 13, 2780–2786 (2009). 70. Christofk, H. R. et al. The M2 splice isoform of
19. Czernin, J. & Phelps, M. E. Positron emission 44. Dang, C. V., Le, A. & Gao, P. MYC‑induced cancer cell pyruvate kinase is important for cancer metabolism
tomography scanning: current and future applications. energy metabolism and therapeutic opportunities. and tumour growth. Nature 452, 230–233 (2008).
Annu. Rev. Med. 53, 89–112 (2002). Clin. Cancer Res. 15, 6479–6483 (2009). The first mechanistic investigation of PKM2 using
20. Le, A. et al. Inhibition of lactate dehydrogenase A 45. Kim, J. W., Gao, P., Liu, Y. C., Semenza, G. L. & Dang, experimental cancer models, confirming the
induces oxidative stress and inhibits tumor C. V. Hypoxia‑inducible factor 1 and dysregulated hypothesis that PKM2 expression provides an
progression. Proc. Natl Acad. Sci. USA 107, c‑Myc cooperatively induce vascular endothelial advantage for tumour growth.
2037–2042 (2010). growth factor and metabolic switches hexokinase 2 71. David, C. J., Chen, M., Assanah, M., Canoll, P. &
21. Fantin, V. R., St-Pierre, J. & Leder, P. Attenuation of and pyruvate dehydrogenase kinase 1. Mol. Cell. Biol. Manley, J. L. HnRNP proteins controlled by c‑Myc
LDH‑A expression uncovers a link between glycolysis, 27, 7381–7393 (2007). deregulate pyruvate kinase mRNA splicing in cancer.
mitochondrial physiology, and tumor maintenance. 46. Dang, C. V., Kim, J. W., Gao, P. & Yustein, J. The Nature 463, 364–368 (2009).
Cancer Cell 9, 425–434 (2006). interplay between MYC and HIF in cancer. Nature Rev. Discovery and explanation of the connection
22. Wong, K. K., Engelman, J. A. & Cantley, L. C. Targeting Cancer 8, 51–56 (2008). between the oncoprotein MYC and PKM2
the PI3K signaling pathway in cancer. Curr. Opin. 47. Li, F. et al. Myc stimulates nuclearly encoded expression.
Genet. Dev. 20, 87–90 (2010). mitochondrial genes and mitochondrial biogenesis. 72. Schneider, J. et al. Tumor M2‑pyruvate kinase in lung
23. Plas, D. R. & Thompson, C. B. Akt‑dependent Mol. Cell. Biol. 25, 6225–6234 (2005). cancer patients: immunohistochemical detection and
transformation: there is more to growth than just 48. Kuhajda, F. P. AMP‑activated protein kinase and disease monitoring. Anticancer Res. 22, 311–318
surviving. Oncogene 24, 7435–7442 (2005). human cancer: cancer metabolism revisited. Int. (2002).
24. Elstrom, R. L. et al. Akt stimulates aerobic glycolysis in J. Obes. 32 (Suppl. 4), S36–S41 (2008). 73. Cerwenka, H. et al. TUM2‑PK (pyruvate kinase type
cancer cells. Cancer Res. 64, 3892–3899 (2004). 49. Shackelford, D. B. & Shaw, R. J. The LKB1‑AMPK tumor M2), CA19–19 and CEA in patients with
25. Fan, Y., Dickman, K. G. & Zong, W. X. Akt and c‑Myc pathway: metabolism and growth control in tumour benign, malignant and metastasizing pancreatic
differentially activate cellular metabolic programs and suppression. Nature Rev. Cancer 9, 563–575 (2009). lesions. Anticancer Res. 19, 849–851 (1999).
prime cells to bioenergetic inhibition. J. Biol. Chem. A comprehensive review of AMPK and LKB1 in 74. Luftner, D. et al. Tumor type M2 pyruvate kinase
285, 7324–7333 (2010). cancer metabolism. expression in advanced breast cancer. Anticancer Res.
26. Robey, R. B. & Hay, N. Is Akt the “Warburg 50. Jones, R. G. et al. AMP‑activated protein kinase 20, 5077–5082 (2000).
kinase”?‑Akt‑energy metabolism interactions and induces a p53‑dependent metabolic checkpoint. Mol. 75. Nathan, C. & Ding, A. SnapShot: reactive oxygen
oncogenesis. Semin. Cancer Biol. 19, 25–31 (2009). Cell 18, 283–293 (2005). intermediates (ROI). Cell 140, 951 (2010).

94 | FebruAry 2011 | vOluMe 11 www.nature.com/reviews/cancer

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

76. Budihardjo, I. I. et al. 6‑Aminonicotinamide sensitizes 100. Ozcan, U. et al. Loss of the tuberous sclerosis complex 125. DeBerardinis, R. J. et al. Beyond aerobic glycolysis:
human tumor cell lines to cisplatin. Clin. Cancer Res. tumor suppressors triggers the unfolded protein transformed cells can engage in glutamine metabolism
4, 117–130 (1998). response to regulate insulin signaling and apoptosis. that exceeds the requirement for protein and
77. Mardis, E. R. et al. Recurring mutations found by Mol. Cell 29, 541–551 (2008). nucleotide synthesis. Proc. Natl Acad. Sci. USA 104,
sequencing an acute myeloid leukemia genome. 101. Nogueira, V. et al. Akt determines replicative 19345–19350 (2007).
N. Engl. J. Med. 361, 1058–1066 (2009). senescence and oxidative or oncogenic premature 126. Gallagher, F. A., Kettunen, M. I., Day, S. E.,
78. Yan, H. et al. IDH1 and IDH2 mutations in gliomas. senescence and sensitizes cells to oxidative apoptosis. Lerche, M. & Brindle, K. M. 13C MR spectroscopy
N. Engl. J. Med. 360, 765–773 (2009). Cancer Cell 14, 458–470 (2008). measurements of glutaminase activity in human
79. Gross, S. et al. Cancer‑associated metabolite 102. Liu, Y. et al. MnSOD inhibits proline oxidase‑induced hepatocellular carcinoma cells using hyperpolarized
2‑hydroxyglutarate accumulates in acute myelogenous apoptosis in colorectal cancer cells. Carcinogenesis 13C‑labeled glutamine. Magn. Reson. Med. 60,
leukemia with isocitrate dehydrogenase 1 and 2 26, 1335–1342 (2005). 253–257 (2008).
mutations. J. Exp. Med. 207, 339–344 (2010). 103. Liu, Y., Borchert, G. L., Surazynski, A. & Phang, J. M. 127. Richards, N. G. & Kilberg, M. S. Asparagine
80. Ward, P. S. et al. The common feature of leukemia‑ Proline oxidase, a p53‑induced gene, targets COX‑2/ synthetase chemotherapy. Annu. Rev. Biochem. 75,
associated IDH1 and IDH2 mutations is a neomorphic PGE2 signaling to induce apoptosis and inhibit tumor 629–654 (2006).
enzyme activity converting α‑ketoglutarate to growth in colorectal cancers. Oncogene 27, 128. Reinert, R. B. et al. Role of glutamine depletion in
2‑hydroxyglutarate. Cancer Cell 17, 225–234 (2010). 6729–6737 (2008). directing tissue‑specific nutrient stress responses to
81. Zhao, S. et al. Glioma‑derived mutations in IDH1 104. Liu, Y. et al. Proline oxidase functions as a mitochondrial L‑asparaginase. J. Biol. Chem. 281, 31222–31233
dominantly inhibit IDH1 catalytic activity and induce tumor suppressor in human cancers. Cancer Res. 69, (2006).
HIF‑1α. Science 324, 261–265 (2009). 6414–6422 (2009). 129. Lunt, S. J., Chaudary, N. & Hill, R. P. The tumor
82. Dang, L. et al. Cancer‑associated IDH1 mutations 105. Budanov, A. V., Sablina, A. A., Feinstein, E., microenvironment and metastatic disease. Clin. Exp.
produce 2‑hydroxyglutarate. Nature 462, 739–744 Koonin, E. V. & Chumakov, P. M. Regeneration of Metastasis 26, 19–34 (2009).
(2009). peroxiredoxins by p53‑regulated sestrins, homologs of 130. Bristow, R. G. & Hill, R. P. Hypoxia and metabolism.
Discovery that driver mutations in IDH1 cause the bacterial AhpD. Science 304, 596–600 (2004). Hypoxia, DNA repair and genetic instability. Nature
acquisition of a novel enzymatic activity and 106. Yoon, K. A., Nakamura, Y. & Arakawa, H. Identification Rev. Cancer 8, 180–192 (2008).
production of 2‑HG. of ALDH4 as a p53‑inducible gene and its protective 131. Semenza, G. L. Regulation of cancer cell metabolism
83. Bleeker, F. E. et al. IDH1 mutations at residue p.R132 role in cellular stresses. J. Hum. Genet. 49, 134–140 by hypoxia‑inducible factor 1. Semin. Cancer Biol. 19,
(IDH1R132) occur frequently in high‑grade gliomas but (2004). 12–16 (2009).
not in other solid tumors. Hum. Mutat. 30, 7–11 107. Suzuki, S. et al. Phosphate‑activated glutaminase 132. Denko, N. C. Hypoxia, HIF1 and glucose metabolism
(2009). (GLS2), a p53‑inducible regulator of glutamine in the solid tumour. Nature Rev. Cancer 8, 705–713
84. Kang, M. R. et al. Mutational analysis of IDH1 codon metabolism and reactive oxygen species. Proc. Natl (2008).
132 in glioblastomas and other common cancers. Int. Acad. Sci. USA 107, 7461–7466 (2010). 133. Wouters, B. G. & Koritzinsky, M. Hypoxia
J. Cancer 125, 353–355 (2009). 108. Chen, W. et al. Direct interaction between Nrf2 and signalling through mTOR and the unfolded protein
85. Giannoni, E., Buricchi, F., Raugei, G., Ramponi, G. & p21Cip1/WAF1 upregulates the Nrf2‑mediated antioxidant response in cancer. Nature Rev. Cancer 8, 851–864
Chiarugi, P. Intracellular reactive oxygen species response. Mol. Cell 34, 663–673 (2009). (2008).
activate Src tyrosine kinase during cell adhesion and 109. Trachootham, D., Alexandre, J. & Huang, P. Targeting 134. Koritzinsky, M. et al. Gene expression during acute
anchorage‑dependent cell growth. Mol. Cell. Biol. 25, cancer cells by ROS‑mediated mechanisms: a radical and prolonged hypoxia is regulated by distinct
6391–6403 (2005). therapeutic approach? Nature Rev. Drug Discov. 8, mechanisms of translational control. EMBO J. 25,
86. Lee, S. R. et al. Reversible inactivation of the tumor 579–591 (2009). 1114–1125 (2006).
suppressor PTEN by H2O2. J. Biol. Chem. 277, 110. Trachootham, D. et al. Effective elimination of 135. Rouschop, K. M. & Wouters, B. G. Regulation of
20336–20342 (2002). fludarabine‑resistant CLL cells by PEITC through a redox‑ autophagy through multiple independent hypoxic
87. Cao, J. et al. Prdx1 inhibits tumorigenesis via mediated mechanism. Blood 112, 1912–1922 (2008). signaling pathways. Curr. Mol. Med. 9, 417–424
regulating PTEN/AKT activity. EMBO J. 28, 111. Trachootham, D. et al. Selective killing of oncogenically (2009).
1505–1517 (2009). transformed cells through a ROS‑mediated 136. Koumenis, C. et al. Regulation of protein synthesis by
88. Gao, P. et al. HIF‑dependent antitumorigenic effect of mechanism by β‑phenylethyl isothiocyanate. Cancer hypoxia via activation of the endoplasmic reticulum
antioxidants in vivo. Cancer Cell 12, 230–238 Cell 10, 241–252 (2006). kinase PERK and phosphorylation of the translation
(2007). 112. Clements, C. M., McNally, R. S., Conti, B. J., Mak, T. W. initiation factor eIF2α. Mol. Cell. Biol. 22, 7405–7416
89. Bell, E. L., Emerling, B. M. & Chandel, N. S. & Ting, J. P. DJ‑1, a cancer‑ and Parkinson’s disease‑ (2002).
Mitochondrial regulation of oxygen sensing. associated protein, stabilizes the antioxidant 137. Bi, M. et al. ER stress‑regulated translation
Mitochondrion 5, 322–332 (2005). transcriptional master regulator Nrf2. Proc. Natl increases tolerance to extreme hypoxia and
90. Ramsey, M. R. & Sharpless, N. E. ROS as a tumour Acad. Sci. USA 103, 15091–15096 (2006). promotes tumor growth. EMBO J. 24, 3470–3481
suppressor? Nature Cell Biol. 8, 1213–1215 113. Gasser, T. et al. Genetic complexity and Parkinson’s (2005).
(2006). disease. Science 277, 388–389 (1997). 138. Romero‑Ramirez, L. et al. XBP1 is essential for
91. Takahashi, A. et al. Mitogenic signalling and the 114. Kim, R. H. et al. Hypersensitivity of DJ‑1‑deficient mice survival under hypoxic conditions and is required
p16INK4a‑Rb pathway cooperate to enforce to 1‑methyl‑4‑phenyl‑1,2,3,6‑tetrahydropyrindine for tumor growth. Cancer Res. 64, 5943–5947
irreversible cellular senescence. Nature Cell Biol. 8, (MPTP) and oxidative stress. Proc. Natl Acad. Sci. USA (2004).
1291–1297 (2006). 102, 5215–5220 (2005). 139. Cairns, R., Papandreou, I. & Denko, N. Overcoming
92. Garrido, C. et al. Mechanisms of cytochrome c release 115. Kim, R. H. et al. DJ‑1, a novel regulator of the tumor physiologic barriers to cancer treatment by
from mitochondria. Cell Death Differ. 13, 1423–1433 suppressor PTEN. Cancer Cell 7, 263–273 (2005). molecularly targeting the tumor microenvironment.
(2006). Discovery that PARK7, a gene mutated in Mol. Cancer Res. 4, 61–70 (2006).
93. Han, D., Antunes, F., Canali, R., Rettori, D. & Parkinson’s disease, is an oncogene. 140. Moreno‑Sanchez, R., Rodriguez‑Enriquez, S.,
Cadenas, E. Voltage‑dependent anion channels 116. Davidson, B. et al. Expression and clinical role of DJ‑1, Marin‑Hernandez, A. & Saavedra, E. Energy
control the release of the superoxide anion from a negative regulator of PTEN, in ovarian carcinoma. metabolism in tumor cells. FEBS J. 274,
mitochondria to cytosol. J. Biol. Chem. 278, Hum. Pathol. 39, 87–95 (2008). 1393–1418 (2007).
5557–5563 (2003). 117. Yuen, H. F. et al. DJ‑1 could predict worse prognosis 141. Yang, C. et al. Glioblastoma cells require glutamate
94. Fruehauf, J. P. & Meyskens, F. L. Reactive oxygen in esophageal squamous cell carcinoma. Cancer dehydrogenase to survive impairments of glucose
species: a breath of life or death? Clin. Cancer Res. Epidemiol. Biomarkers Prev. 17, 3593–3602 (2008). metabolism or Akt signaling. Cancer Res. 69,
13, 789–794 (2007). 118. Lin, M. T. & Beal, M. F. Mitochondrial dysfunction and 7986–7993 (2009).
95. Bae, Y. S. et al. Epidermal growth factor (EGF)‑induced oxidative stress in neurodegenerative diseases. Nature 142. Sonveaux, P. et al. Targeting lactate‑fueled respiration
generation of hydrogen peroxide. Role in EGF 443, 787–795 (2006). selectively kills hypoxic tumor cells in mice. J. Clin.
receptor‑mediated tyrosine phosphorylation. J. Biol. 119. Bajaj, A., Driver, J. A. & Schernhammer, E. S. Invest. 118, 3930–3942 (2008).
Chem. 272, 217–221 (1997). Parkinson’s disease and cancer risk: a systematic
96. Sundaresan, M., Yu, Z. X., Ferrans, V. J., Irani, K. & review and meta‑analysis. Cancer Causes Control 21, Acknowledgements
Finkel, T. Requirement for generation of H2O2 for 697–707 (2010). The authors would like to thank M. Saunders for scientific
platelet‑derived growth factor signal transduction. 120. Coles, N. W. & Johnstone, R. M. Glutamine editing of the Review.
Science 270, 296–299 (1995). metabolism in Ehrlich ascites‑carcinoma cells.
97. Vaughn, A. E. & Deshmukh, M. Glucose metabolism Biochem. J. 83, 284–291 (1962). Competing interests statement
inhibits apoptosis in neurons and cancer cells by redox 121. Reitzer, L. J., Wice, B. M. & Kennell, D. Evidence that The authors declare no competing financial interests.
inactivation of cytochrome c. Nature Cell Biol. 10, glutamine, not sugar, is the major energy source for
1477–1483 (2008). cultured HeLa cells. J. Biol. Chem. 254, 2669–2676
Evidence that redox control by the GSH system is (1979). DATABASES
important in neurons and cancer cells and that 122. Eagle, H. Nutrition needs of mammalian cells in tissue National Cancer Institute Drug Dictionary:
reduction of cytochrome c prevents apoptosis. culture. Science 122, 501–514 (1955). http://www.cancer.gov/drugdictionary
98. Schafer, Z. T. et al. Antioxidant and oncogene rescue 123. Wise, D. R. et al. Myc regulates a transcriptional l-asparaginase
of metabolic defects caused by loss of matrix program that stimulates mitochondrial glutaminolysis Pathway Interaction Database: http://pid.nci.nih.gov
attachment. Nature 461, 109–113 (2009). and leads to glutamine addiction. Proc. Natl Acad. Sci. AMPK | HIF1 | HIF2 | LKB1 | mTOR | MYC | p53 | PI3K
99. Li, B., Gordon, G. M., Du, C. H., Xu, J. & Du, W. USA 105, 18782–18787 (2008).
Specific killing of Rb mutant cancer cells by 124. Gao, P. et al. c‑Myc suppression of miR‑23a/b enhances
FURTHER INFORMATION
Tak W. Mak’s homepage:
inactivating TSC2. Cancer Cell 17, 469–480 (2010). mitochondrial glutaminase expression and glutamine
http://medbio.utoronto.ca/faculty/mak.html
Evidence that inappropriate activation of growth metabolism. Nature 458, 762–765 (2009).
and proliferation pathways can lead to excessive Strong mechanistic evidence that MYC participates all lInKS are aCtIve In tHe OnlIne PDf
stress and cell death. in promoting mitochondrial glutaminase activity.

nATure revIeWs | CanCer vOluMe 11 | FebruAry 2011 | 95

© 2011 Macmillan Publishers Limited. All rights reserved

You might also like